Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Gynecol Endocrinol ; 39(1): 2269281, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37844908

RESUMEN

OBJECTIVE: This study is aimed to determine the efficacy of a cocktail style treatment by combining GnRH-antagonist, letrozole, and mifepristone on the prevention of ovarian hyperstimulation syndrome (OHSS) in high-risk women. METHODS: This prospective, randomized controlled clinical trial was performed between January 2018 and December 2018. A total of 170 women who identified as high risk of OHSS during the ovarian hyperstimulation and underwent cryopreservation of whole embryos. On the day of oocyte retrieval, the combination group received 0.25 mg Cetrorelix for 3 d, 5 mg letrozole for 5 d, and 50 mg mifepristone for 3 d, the mifepristone group received 50 mg mifepristone for 3 d. A total of 156 cases were included in final analysis. All the frozen embryo transfer (FET) cycles were followed up until December 2021. RESULTS: The combination group showed significantly decreased incidence of moderate and severe OHSS than mifepristone group (20.5% vs. 42.3%), with remarkably reduced serum estradiol level on hCG + 3 and + 5 d, decreased ovarian diameter, and shortened luteal phase. Oocyte retrieval number, levels of estradiol on hCG + 0 and VEGF, and ovarian diameter on hCG + 5 were associated with the severity of the symptoms. There was no significant difference in cumulative live birth rates (LBRs) between the combination and mifepristone group (74.4% vs. 76.9%). CONCLUSIONS: The combination treatment effectively reduces the incidence of moderate/severe OHSS in high-risk women.


Asunto(s)
Síndrome de Hiperestimulación Ovárica , Femenino , Humanos , Síndrome de Hiperestimulación Ovárica/complicaciones , Letrozol/uso terapéutico , Mifepristona , Fertilización In Vitro , Estudios Prospectivos , Estradiol , Hormona Liberadora de Gonadotropina , Antagonistas de Hormonas/uso terapéutico , Inducción de la Ovulación/efectos adversos
2.
Reprod Biomed Online ; 42(2): 291-300, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33249057

RESUMEN

RESEARCH QUESTION: Can luteolysis-targeted drugs, gonadotrophin-releasing hormone antagonist (GnRH-ant), mifepristone and letrozole, administered separately or in combination, prevent the progression of ovarian hyperstimulation syndrome (OHSS) in a rat model? DESIGN: Thirty-six female Wistar rats were randomly divided into six groups, including control group (OHSS group, ovarian hyperstimulation-induced OHSS); GnRH-ant group (OHSS with GnRH-ant treatment); mifepristone group (OHSS with mifepristone treatment); letrozole group (OHSS with letrozole treatment); combination group (OHSS with GnRH-ant, mifepristone and letrozole treatment in combination). The main outcomes were the alterations in OHSS-related indices, including ovarian weight, vascular permeability, serum oestradiol and progesterone levels, corpus luteum proportion and diameter, ovarian vascular endothelial growth factor (VEGF), interleukin 6 (IL-6), caspase-3 and cleaved caspase-3 levels. RESULTS: No significant difference was found in body weight gain among the six groups. Compared with the control group, the OHSS group showed significant increases in all OHSS-related indices. GnRH-ant treatment showed decreases in vascular permeability, serum oestradiol level, corpus luteum diameter, ovarian VEGF /IL-6 mRNA levels, and increases in ovarian caspase-3 and cleaved caspase-3 levels. Mifepristone treatment demonstrated reduction in serum progesterone level and corpus luteum diameter, and elevation in ovarian caspase-3 and cleaved caspase-3 levels. Letrozole treatment displayed a decline in serum oestradiol level and corpus luteum diameter, and up-regulation in ovarian caspase-3 and cleaved caspase-3 levels. The combination treatment by GnRH-ant, mifepristone and letrozole showed enhanced synergistic effect on reducing OHSS-related indices. CONCLUSIONS: GnRH-ant, mifepristone and letrozole are beneficial in preventing the progression of OHSS through different luteolytic mechanisms. Cocktail style treatment shows enhanced synergistic effect on preventing the progression of OHSS.


Asunto(s)
Inhibidores de la Aromatasa/uso terapéutico , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Letrozol/uso terapéutico , Mifepristona/uso terapéutico , Síndrome de Hiperestimulación Ovárica/prevención & control , Progesterona/antagonistas & inhibidores , Animales , Caspasa 3/metabolismo , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Interleucina-6/metabolismo , Ovario/efectos de los fármacos , Ovario/metabolismo , Distribución Aleatoria , Ratas Wistar , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Med Sci Monit ; 27: e927709, 2021 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-33390585

RESUMEN

BACKGROUND This study aims to explore the effect of Sinomenine (SIN) on pregnancy outcomes of recurrent spontaneous abortion (RSA) in a mouse model. MATERIAL AND METHODS Thirty female CBA/J mice were allocated into 3 groups randomly, then mated with BALB/c mice (CBA/J×BALB/c) as normal-pregnancy group (n=10), or mated with DBA/2 mice (CBA/J×DBA/2) as RSA model (n=10), or CBA/J×DBA/2 mice treated with SIN as RSA+SIN group (n=10). The number of surviving and reabsorbed embryos in each group were counted on day 13.5 of gestation. The mouse serum was collected to determine the levels of interferon-γ (IFN)-γ and IL-4 by ELISA. Immunohistochemistry, qRT-PCR and immunoblotting were used to determine the location, mRNA and protein expressions of IFN-γ, IL-4, T-bet and GATA3 in the decidual and placental tissue. RESULTS In the RSA group, the amount of reabsorbed embryo was significantly higher than that in the normal-pregnancy group. However, SIN treatment showed a rescue effect on spontaneous abortion in RSA mice. IFN-γ, IL-4, T-bet, and GATA3 were all expressed in placental tissues and mainly located in the cytoplasm. The RSA group demonstrated higher expression levels of IFN-γ and T-bet than in the RSA+SIN and normal-pregnancy groups. Although RSA and RSA+SIN groups showed lower expression levels of IL-4 and GATA3 than in the normal-pregnancy group, there was no significant difference between RSA and RSA+SIN groups regarding IL-4 and GATA expression levels. CONCLUSIONS SIN treatment demonstrates a therapeutic effect on spontaneous abortion in RSA mice, possibly through regulating the balance of Th1/Th2 in maternal circulation and decidual tissues.


Asunto(s)
Aborto Espontáneo/tratamiento farmacológico , Aborto Espontáneo/inmunología , Embrión de Mamíferos/patología , Morfinanos/uso terapéutico , Balance Th1 - Th2/efectos de los fármacos , Aborto Espontáneo/sangre , Animales , Decidua/metabolismo , Modelos Animales de Enfermedad , Embrión de Mamíferos/efectos de los fármacos , Femenino , Factor de Transcripción GATA3/metabolismo , Interferón gamma/sangre , Interleucina-4/sangre , Masculino , Ratones , Morfinanos/farmacología , Placenta/metabolismo , Embarazo , Proteínas de Dominio T Box/metabolismo
4.
Int J Mol Sci ; 21(12)2020 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-32570961

RESUMEN

Normal pregnancy is associated with dramatic increases in uterine blood flow to facilitate the bidirectional maternal-fetal exchanges of respiratory gases and to provide sole nutrient support for fetal growth and survival. The mechanism(s) underlying pregnancy-associated uterine vasodilation remain incompletely understood, but this is associated with elevated estrogens, which stimulate specific estrogen receptor (ER)-dependent vasodilator production in the uterine artery (UA). The classical ERs (ERα and ERß) and the plasma-bound G protein-coupled ER (GPR30/GPER) are expressed in UA endothelial cells and smooth muscle cells, mediating the vasodilatory effects of estrogens through genomic and/or nongenomic pathways that are likely epigenetically modified. The activation of these three ERs by estrogens enhances the endothelial production of nitric oxide (NO), which has been shown to play a key role in uterine vasodilation during pregnancy. However, the local blockade of NO biosynthesis only partially attenuates estrogen-induced and pregnancy-associated uterine vasodilation, suggesting that mechanisms other than NO exist to mediate uterine vasodilation. In this review, we summarize the literature on the role of NO in ER-mediated mechanisms controlling estrogen-induced and pregnancy-associated uterine vasodilation and our recent work on a "new" UA vasodilator hydrogen sulfide (H2S) that has dramatically changed our view of how estrogens regulate uterine vasodilation in pregnancy.


Asunto(s)
Estrógenos/metabolismo , Receptores de Estrógenos/metabolismo , Arteria Uterina/patología , Epigénesis Genética , Femenino , Humanos , Óxido Nítrico/metabolismo , Embarazo , Receptores de Estrógenos/genética , Arteria Uterina/metabolismo , Vasodilatación
5.
J Cell Physiol ; 234(6): 9264-9273, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30317617

RESUMEN

Endogenous hydrogen sulfide (H2 S), synthesized by cystathionine ß-synthase (CBS) and cystathionine γ-lyase (CSE), is a potent vasodilator that can be stimulated by estradiol-17ß (E 2 ß) in uterine artery (UA) smooth muscle (UASMC) in vivo; however, the underlying mechanisms are unknown. This study tested a hypothesis that E 2 ß stimulates H 2 S biosynthesis by upregulating CBS expression via specific estrogen receptor (ER). Treatment with E 2 ß stimulated time- and concentration- dependent CBS and CSE messenger RNA (mRNA) and protein expressions, and H 2 S production in cultured primary UASMC isolated from late pregnant ewes, which were blocked by ICI 182,780. Treatment with specific ERα or ERß agonist mimicked these E 2 ß-stimulated responses, which were blocked by specific ERα or ERß antagonist. Moreover, E 2 ß activated both CBS and CSE promoters and ICI 182,780 blocked the E 2 ß-stimulated responses. Thus, E 2 ß stimulates H 2 S production by upregulating CBS and CSE expression via specific ER-dependent transcription in UASMC in vitro.


Asunto(s)
Cistationina betasintasa/metabolismo , Cistationina gamma-Liasa/metabolismo , Estradiol/farmacología , Sulfuro de Hidrógeno/metabolismo , Miocitos del Músculo Liso/metabolismo , Receptores de Estrógenos/metabolismo , Transcripción Genética/efectos de los fármacos , Arteria Uterina/citología , Animales , Células Cultivadas , Cistationina betasintasa/genética , Cistationina gamma-Liasa/genética , Miocitos del Músculo Liso/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ovinos
6.
Biol Reprod ; 100(6): 1630-1636, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30772913

RESUMEN

Uterine vasodilation dramatically increases during the follicular phase of the estrous cycle and pregnancy, which are estrogen-dominant physiological states. Uterine vasodilation is believed to be mainly controlled by local uterine artery (UA) production of vasodilators and angiogenic factors. The extremely potent vasodilator and proangiogenic hydrogen sulfide (H2S) is synthesized via metabolizing L-cysteine by cystathionine ß-synthase (CBS) and cystathionine γ-lyase (CTH). This study was designed to determine if UA H2S production increases with augmented expression and/or activity of CBS and/or CTH during the ovarian cycle and pregnancy in sheep. Uterine arteries from intact nonpregnant (NP) luteal and follicular phase and late (130-135 days, term ≈ 145 days) pregnant (P) ewes were collected; endothelium-enriched proteins (UAendo) and endothelium-denuded smooth muscle (UAvsm) were mechanically prepared for accessing CBS and CTH proteins by immunoblotting; their cellular localization was determined by semi-quantitative immunofluorescence microscopy. H2S production was measured by the methylene blue assay. Immunoblotting revealed that CBS but not CTH protein was greater in P > > > NP follicular > luteal UAendo and UAvsm (P < 0.001). H2S production was greater in P > > > NP UAendo and UAvsm (P < 0.01). Pregnancy-augmented UAendo and UAvsm H2S production was inhibited by the specific CBS but not CTH inhibitor. CBS and CTH proteins were localized to both endothelium and smooth muscle; however, only CBS protein was significantly greater in P vs NP UA endothelium and smooth muscle. Thus, ovine UA H2S production is significantly augmented via selectively upregulating endothelium and smooth muscle CBS during the follicular phase and pregnancy in vivo.


Asunto(s)
Ciclo Estral/fisiología , Sulfuro de Hidrógeno/metabolismo , Preñez , Ovinos/metabolismo , Arteria Uterina/metabolismo , Animales , Femenino , Edad Gestacional , Ovario/fisiología , Embarazo , Preñez/fisiología , Vasodilatación/fisiología
7.
Biol Reprod ; 100(2): 514-522, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30277497

RESUMEN

Endogenous hydrogen sulfide (H2S) is a potent vasodilator and proangiogenic second messenger synthesized from L-cysteine by cystathionine ß-synthase (CBS) and cystathionine γ-lyase (CTH). Estrogens are potent vasodilators that stimulate H2S biosynthesis in uterine arteries (UA) in vivo; however, the underlying mechanisms are unknown. We hypothesized that estrogens stimulate H2S biosynthesis in UA endothelial cells (UAEC) via specific estrogen receptor (ER)-dependent mechanisms. In cultured primary UAEC, treatment with estradiol-17ß (E2ß) stimulated CBS and CTH mRNAs and proteins in a time- and concentration-dependent fashion. As little as 0.1 nM E2ß was effective in increasing CBS and CTH expressions and these stimulatory effects maximized with 10-100 nM E2ß at 48-72 h. E2ß also activated CBS and CTH promoters in UAEC, leading to CBS and CTH expression. Treatment with E2ß stimulated H2S production, which was blocked by specific inhibitors of either CBS or CTH and their combination and the ER antagonist ICI 182780. Treatment with either specific agonist of ERα or ERß stimulated both CBS and CTH mRNA and protein expressions and H2S production to levels similar to that of E2ß. Specific antagonist of either ERα or ERß blocked E2ß-stimulated CBS and CTH mRNA and protein expressions and H2S production. Combinations of either ERα or ERß agonists or their antagonists had no additive effects. Thus, E2ß stimulates H2S production by upregulating CBS and CTH mRNA and protein expressions through specific ERα or ERß-dependent CBS and CTH transcription in UAEC in vitro.


Asunto(s)
Cistationina betasintasa/metabolismo , Cistationina gamma-Liasa/metabolismo , Células Endoteliales/efectos de los fármacos , Estradiol/farmacología , Sulfuro de Hidrógeno/metabolismo , Ovinos , Animales , Cistationina betasintasa/genética , Cistationina gamma-Liasa/genética , Relación Dosis-Respuesta a Droga , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos , Arteria Uterina/citología , Útero/irrigación sanguínea
8.
Reproduction ; 156(6): 569-577, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30328351

RESUMEN

Recent studies revealed that TMEM16A is involved in several reproductive processes, including ovarian estrogen secretion and ovulation, sperm motility and acrosome reaction, fertilization and myometrium contraction. However, little is known about the expression and function of TMEM16A in embryo implantation and decidualization. In this study, we focused on the expression and regulation of TMEM16A in mouse uterus during early pregnancy. We found that TMEM16A is upregulated in uterine endometrium in response to embryo implantation and decidualization. Progesterone treatment could induce TMEM16A expression in endometrial stromal cells through progesterone receptor/c-Myc pathway, which is blocked by progesterone receptor antagonist or the inhibitor of c-Myc signaling pathway. Inhibition of TMEM16A by small molecule inhibitor (T16Ainh-A01) resulted in impaired embryo implantation and decidualization in mice. Treatment with either specific siRNA of Tmem16a or T16Ainh-A01 inhibited the decidualization and proliferation of mouse endometrial stromal cells. In conclusion, our results revealed that TMEM16A is involved in embryo implantation and decidualization in mice, compromised function of TMEM16A may lead to impaired embryo implantation and decidualization.


Asunto(s)
Anoctamina-1/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Implantación del Embrión/efectos de los fármacos , Pirimidinas/farmacología , Células del Estroma/efectos de los fármacos , Tiazoles/farmacología , Útero/efectos de los fármacos , Animales , Anoctamina-1/genética , Anoctamina-1/metabolismo , Células Cultivadas , Decidua/efectos de los fármacos , Decidua/metabolismo , Femenino , Ratones , Embarazo , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Células del Estroma/metabolismo , Útero/metabolismo
9.
Reproduction ; 155(5): 423-432, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29420252

RESUMEN

OPN is essential for blastocyst implantation and placentation. Previous study found that miR181a was increased while miR181b was downregulated in endometrium during decidualization. However, the information regarding their effects on decidualization in human endometrium is still limited. Here, we report a novel role of OPN and miR181b in uterine decidualization and pregnancy success in humans. The expression of OPN was high in endometrium in secretory phase and in vitro decidualized hESC, whereas miR181b expression was low in identical conditions. Further analysis confirmed that OPN expression was upregulated by cAMP and C/EBPß signal pathway, while downregulated by miR181b. Increased OPN expression could promote the expression of decidualization-related and angiogenesis-related genes. Conversely, the processes of decidualization and angiogenesis in hESC were compromised by inhibiting OPN expression in vitro OPN expression was repressed in implantation failure group when compared with successful pregnancy group in IVF/ICSI-ET cycles. These findings add a new line of evidence supporting the fact that OPN is involved in decidualization and pregnancy success.


Asunto(s)
Implantación del Embrión/fisiología , Endometrio/metabolismo , Osteopontina/metabolismo , Células del Estroma/metabolismo , Línea Celular , Decidua/citología , Decidua/metabolismo , Endometrio/citología , Femenino , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Osteopontina/genética , Embarazo , Transducción de Señal/fisiología , Células del Estroma/citología , Regulación hacia Arriba
10.
J Biol Chem ; 290(35): 21280-91, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26178372

RESUMEN

Decidualization is an essential process of maternal endometrial stromal cells to support pregnancy. Although it is known that enhanced glucose influx is critical for decidualization, the underlying mechanism in regulating glucose metabolism in decidua remains insufficiently understood. Here, we demonstrate that aerobic glycolysis-related genes and factors are all substantially induced during decidualization, indicating the existence of Warburg-like glycolysis in decidua. In vitro, progesterone activates hypoxia-inducible factor 1α (Hif1α) and c-Myc through Pi3k-Akt signaling pathway to maintain aerobic glycolysis in decidualizing cells. Knocking down of pyruvate kinase M2 (Pkm2) attenuates the induction of decidual marker gene. Decidual formation in vivo is also impaired by glycolysis inhibitor 3-bromopyruvate. Besides, lactate exporter monocarboxylate transporter 4 (Mct4) is induced in newly formed decidual cells, whereas lactate importer Mct1 and proliferation marker Ki-67 are complementarily located in the surrounding undifferentiated cells, which are supposed to consume lactate for proliferation. Hif1α activation is required for lactate-dependent proliferation of the undifferentiated cells. Inhibition of lactate flux leads to compromised decidualization and decelerated lactate-dependent proliferation. In summary, we reveal that Warburg-like glycolysis and local lactate shuttle are activated in decidua and play important roles for supporting early pregnancy.


Asunto(s)
Endometrio/citología , Glucólisis , Ácido Láctico/metabolismo , Ratones/fisiología , Preñez/fisiología , Animales , Células Cultivadas , Endometrio/fisiología , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Embarazo , Progesterona/metabolismo , Células del Estroma/citología , Células del Estroma/metabolismo
11.
J Biol Chem ; 287(19): 15174-92, 2012 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-22403396

RESUMEN

Embryo implantation into the maternal uterus is a crucial step for the successful establishment of mammalian pregnancy. Following the attachment of embryo to the uterine luminal epithelium, uterine stromal cells undergo steroid hormone-dependent decidualization, which is characterized by stromal cell proliferation and differentiation. The mechanisms underlying steroid hormone-induced stromal cell proliferation and differentiation during decidualization are still poorly understood. Ribonucleotide reductase, consisting of two subunits (RRM1 and RRM2), is a rate-limiting enzyme in deoxynucleotide production for DNA synthesis and plays an important role in cell proliferation and tumorgenicity. Based on our microarray analysis, Rrm2 expression was significantly higher at implantation sites compared with interimplantation sites in mouse uterus. However, the expression, regulation, and function of RRM2 in mouse uterus during embryo implantation and decidualization are still unknown. Here we show that although both RRM1 and RRM2 expression are markedly induced in mouse uterine stromal cells undergoing decidualization, only RRM2 is regulated by progesterone, a key regulator of decidualization. Further studies showed that the induction of progesterone on RRM2 expression in stromal cells is mediated by the AKT/c-MYC pathway. RRM2 can also be induced by replication stress and DNA damage during decidualization through the ATR/ATM-CHK1-E2F1 pathway. The weight of implantation sites and deciduoma was effectively reduced by specific inhibitors for RRM2. The expression of decidual/trophoblast prolactin-related protein (Dtprp), a reliable marker for decidualization in mice, was significantly reduced in deciduoma and steroid-induced decidual cells after HU treatment. Therefore, RRM2 may be an important effector of progesterone signaling to induce cell proliferation and decidualization in mouse uterus.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Daño del ADN , Progesterona/farmacología , Ribonucleósido Difosfato Reductasa/genética , Útero/efectos de los fármacos , Animales , Western Blotting , Células Cultivadas , Decidua/citología , Decidua/efectos de los fármacos , Decidua/metabolismo , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F1/metabolismo , Implantación del Embrión/efectos de los fármacos , Implantación del Embrión/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hidroxiurea/farmacología , Masculino , Ratones , Ovariectomía , Embarazo , Unión Proteica , Proteínas Proto-Oncogénicas c-myc/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleósido Difosfato Reductasa/metabolismo , Ribonucleótido Reductasas/genética , Ribonucleótido Reductasas/metabolismo , Transducción de Señal/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Factores de Tiempo , Útero/citología , Útero/metabolismo
12.
Int J Mol Sci ; 14(7): 14504-17, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23852023

RESUMEN

Embryo implantation is a highly synchronized bioprocess between an activated blastocyst and a receptive uterus. In mice, successful implantation relies on the dynamic interplay of estrogen and progesterone; however, the key mediators downstream of these hormones that act on blastocyst competency and endometrium receptivity acquisition are largely unknown. In this study, we showed that the expression of osteopontin (OPN) in mouse blastocysts is regulated by ovarian estrogen and uterine micro-environment. OPN mRNA is up-regulated in mouse blastocyst on day 4 of pregnancy, which is associated with ovarian estrogen secretion peak. Hormone treatment in vivo demonstrated that OPN expression in a blastocyst is regulated by estrogen through an estrogen receptor (ER). Our results of the delayed and activated implantation model showed that OPN expression is induced after estrogen injection. While estrogen treatment during embryo culture in vitro showed less effect on OPN expression, the tubal ligation model on day 3 of pregnancy confirmed that the regulation of estrogen on OPN expression in blastocyst might, through some specific cytokines, have existed in a uterine micro-environment. Collectively, our study presents that estrogen regulates OPN expression and it may play an important role during embryo implantation by activating blastocyst competence and facilitating the endometrium acceptable for active blastocyst.


Asunto(s)
Blastocisto/metabolismo , Estrógenos/farmacología , Osteopontina/metabolismo , Útero/metabolismo , Animales , Citocinas/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Estrógenos/metabolismo , Femenino , Ratones , Osteopontina/genética , Embarazo , ARN Mensajero/metabolismo , Receptores de Estrógenos/metabolismo , Regulación hacia Arriba
13.
J Clin Med ; 12(5)2023 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-36902791

RESUMEN

This study aims to analyze the cycle characteristics, pregnancy, and neonatal outcomes in early rescue intracytoplasmic sperm injection (r-ICSI) cycles in normal and hyper-ovarian response women in their first IVF/ICSI attempts. Data from short-term in vitro fertilization (IVF, N = 7148), early r-ICSI (N = 618), and ICSI (N = 1744) cycles were retrospectively analyzed from normal and hyper-ovarian women who underwent their first IVF/ICSI cycles at our center from October 2015 to October 2021. The r-ICSI group was subdivided into partial r-ICSI (N = 451) and total r-ICSI (N = 167) based on the number of fertilized oocytes in the IVF part. Cyclic characteristics, pregnancy, delivery and neonatal outcomes in the fresh cycle were compared among the four groups; pregnancy, delivery and neonatal outcomes in frozen-thawed cycles were compared regarding cleavage and blastocyst transfers derived from r-ICSI cycles. Partial r-ICSI cycles showed different cyclic characteristics compared to total r-ICSI cycles, presenting as elevated AMH and estradiol levels on trigger day and an increased number of oocytes retrieved. Early r-ICSI delayed blastocyst development as seen by the increase in the number of day 6 blastocysts. There was no significant difference among the groups in clinical pregnancy, pregnancy loss, and live birth in fresh cleavage-stage embryo transfer cycles. However, early r-ICSI groups showed a reduction in clinical pregnancy and live birth rates in fresh blastocyst transfer cycles but not in the frozen-thawed cycles. For pregnant women, early r-ICSI did not show a negative effect on the risk of preterm birth, Cesarean section, neonatal birth weight, and sex ratio. In conclusion, early r-ICSI had comparable pregnancy, delivery, and neonatal outcomes when compared with short-term IVF and ICSI groups in fresh cleavage-stage embryo transfer cycles, but early r-ICSI did result in reduced pregnancy outcomes in fresh blastocyst embryo cycles, possibly due to delayed blastocyst development and asynchronization with the endometrium.

14.
Biology (Basel) ; 10(5)2021 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-34068010

RESUMEN

A cutaneous squamous cell carcinoma (cSCC) derived from keratinocytes is the second most common cause of non-melanoma skin cancer. The accumulation of the mutational burden of genes and cellular DNA damage caused by the risk factors (e.g., exposure to ultraviolet radiation) contribute to the aberrant proliferation of keratinocytes and the formation of a cSCC. A cSCC encompasses a spectrum of diseases that range from recursor actinic keratosis (AK) and squamous cell carcinoma (SCC) in situ (SCCIS) to invasive cSCCs and further metastatic SCCs. Emerging evidence has revealed that lncRNAs are involved in the biological process of a cSCC. According to the ceRNA regulatory theory, lncRNAs act as natural miRNA sponges and interact with miRNA response elements, thereby regulating the mRNA expression of their down-stream targets. This study was designed to search for the potential lncRNAs that may become potential therapeutic targets or biomarkers of a cSCC. Considering the spirit of the study to be adequately justified, we collected microarray-based datasets of 19 cSCC tissues and 12 normal skin samples from the GEO database (GSE42677 and GSE45164). After screening the differentially expressed genes via a limma package, we identified 24 differentially expressed lncRNAs (DElncRNAs) and 3221 differentially expressed mRNAs (DEmRNAs). The miRcode, miRTarBase, miRDB and TargetScan databases were used to predict miRNAs that could interact with DElncRNAs and DEmRNAs. A total of 137 miRNA-lncRNA and 221 miRNA-mRNA pairs were retained in the ceRNA network, consisting of 31 miRNAs, 11 DElncRNAs and 155 DEmRNAs. For the functional analysis, the top enriched biological process was enhancer sequence-specific DNA binding in Gene Ontology (GO) terms. The FoxO signaling pathway, autophagy and cellular senescence were the top enrichment terms based on a Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. The combination of a STRING tool and Cytoscape software (plug-in MCODE) identified five core mRNAs and built a core mRNA-associated ceRNA network. The expression for five identified core mRNAs and their related nine lncRNAs was validated using the external dataset GSE7553. Finally, one lncRNA HLA-F-AS1 and three mRNAs named AGO4, E2F1 and CCND1 were validated with the same expression patterns. We speculate that lncRNA HLA-F-AS1 may sponge miR-17-5p or miR-20b-5p to regulate the expression of CCND1 and E2F1 in the cSCC. The present study may provide potential diagnostic and therapeutic targets for cSCC patients.

15.
J Int Med Res ; 48(6): 300060520918474, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32586174

RESUMEN

OBJECTIVE: This study aimed to compare the pregnancy outcomes between women receiving frozen embryo transfer (FET) with hormone replacement treatment (HRT) with and without gonadotropin-releasing hormone agonist (GnRHa) pretreatment. METHODS: All consecutive women undergoing HRT cycles (2936 cycles) or HRT with GnRHa pretreatment (HRT + GnRHa, 303 cycles) at our reproductive center between January 2015 and December 2017 were analyzed retrospectively. RESULTS: The average age was higher in the HRT + GnRHa compared with the HRT group (34.0 ± 4.8 vs. 31.3 ± 4.4). However, the pregnancy outcomes were comparable between the two groups. The clinical pregnancy rate was significantly increased in younger women (≤35 years) in the HRT + GnRHa group compared with the HRT group (56.8% vs. 48.7%), but the live birth rates were similar in the two groups (44.2% vs. 38.4%). The HRT + GnRHa protocol significantly increased the clinical pregnancy rate (55.6% vs. 43.2%) and live birth rate (43.5% vs. 33.5%) compared with the HRT group among women with endometriosis, and significantly decreased the abortion rate in women with polycystic ovarian syndrome (3.1% vs. 16.4%). CONCLUSIONS: GnRHa pretreatment may improve pregnancy outcomes in women with endometriosis and polycystic ovarian syndrome.


Asunto(s)
Transferencia de Embrión , Hormona Liberadora de Gonadotropina , Femenino , Fertilización In Vitro , Terapia de Reemplazo de Hormonas , Humanos , Inducción de la Ovulación , Embarazo , Índice de Embarazo , Estudios Retrospectivos
16.
Life Sci ; 260: 118439, 2020 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-32950574

RESUMEN

AIMS: This study aims to investigate the effects of intrauterine perfusion of granulocyte colony-stimulating factor (G-CSF) on a thin-endometrium rat model. MAIN METHODS: Twenty rats in two groups of 10 were used. Group I was perfused with normal saline (NS) in the right uterine horn and 95% ethanol in the left one. Group II was bilaterally perfused with 95% ethanol into the uterine horns. After three estrous cycles, Group II was perfused with NS in the right uterine horn and G-CSF (30 µg/kg) in the left one. Hematoxylin-eosin (HE) and immunohistochemistry (IHC) staining were used to detect changes in endometrial thickness and expression of cytokeratin 19 (CK19) and vimentin (Vim). The relative expression levels of vascular endothelial growth factor (Vegf) and leukemia inhibitory factor (Lif) were also tested via reverse transcription-quantitative real-time polymerase chain reaction (RT-qPCR) and Western-blot analyses. KEY FINDINGS: G-CSF treatment significantly increased the thickness of the endometrium in the 95% ethanol-induced thin-endometrium rat model. The expression levels of endometrial glandular epithelial cell marker for CK19 and stromal cell marker Vim were augmented in the G-CSF-treated group compared with the control group. Moreover, G-CSF treatment stimulated the expression of VEGF and LIF in the 95% ethanol-induced thin-endometrium rat model. SIGNIFICANCE: G-CSF intrauterine perfusion improved endometrial receptivity in the thin-endometrium rat model by stimulating endometrial proliferation and angiogenesis.


Asunto(s)
Endometrio/efectos de los fármacos , Endometrio/fisiopatología , Factor Estimulante de Colonias de Granulocitos/farmacología , Animales , Endometrio/fisiología , Etanol/toxicidad , Femenino , Expresión Génica/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Factor Inhibidor de Leucemia/genética , Perfusión , Ratas Sprague-Dawley , Útero/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , Vimentina/metabolismo
17.
Endocrinology ; 161(11)2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32987401

RESUMEN

Angiogenesis is a physiological process for endometrial regeneration in the menstrual cycle and remodeling during pregnancy. Endogenous hydrogen sulfide (H2S), produced by cystathionine-ß synthase (CBS) and cystathionine-γ lyase (CSE), is a potent proangiogenic factor; yet, whether the H2S system is expressed in the endometrium and whether H2S plays a role in endometrial angiogenesis are unknown. This study was to test whether estrogens stimulate endometrial H2S biosynthesis to promote endometrial microvascular endothelial cell (EMEC) angiogenesis. CBS messenger RNA/protein and H2S production significantly differed among endometria from postmenopausal (POM), premenopausal secretory (sPRM), and proliferative (pPRM) nonpregnant (NP) and pregnant (Preg) women (P < .05) in a rank order of POM approximately equal to sPRM is less than pPRM is less than Preg, positively correlating with angiogenesis indices and endogenous estrogens and with no difference in CSE expression. CBS and CSE proteins were localized to stroma, glands, and vessels in endometrium, and greater stromal CBS protein was observed in the pPRM and Preg states. Estradiol-17ß (E2) (but not progesterone) stimulated CBS (but not CSE) expression and H2S production in pPRM endometrial stromal cells (ESCs) in vitro, which were attenuated by ICI 182 780. The H2S donor sodium hydrosulfide promoted in vitro EMEC angiogenesis. Co-culture with sPRM, pPRM, and Preg ESCs all stimulated EMEC migration with a rank order of sPRM less than pPRM approximately equal to Preg. CBS (but not CSE) inhibition attenuated ESC-stimulated EMEC migration. E2 did not affect EMEC migration but potentiated ESC-stimulated EMEC migration. Altogether, estrogens stimulate specific receptor-dependent stromal CBS-H2S production to promote endometrial EMEC angiogenesis in women.


Asunto(s)
Cistationina betasintasa/metabolismo , Endometrio/efectos de los fármacos , Estradiol/farmacología , Sulfuro de Hidrógeno/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Células del Estroma/metabolismo , Adulto , Anciano , Inductores de la Angiogénesis/metabolismo , Células Cultivadas , Endometrio/irrigación sanguínea , Endometrio/citología , Endometrio/metabolismo , Femenino , Humanos , Ciclo Menstrual/efectos de los fármacos , Ciclo Menstrual/genética , Ciclo Menstrual/metabolismo , Persona de Mediana Edad , Neovascularización Fisiológica/genética , Posmenopausia/efectos de los fármacos , Posmenopausia/genética , Posmenopausia/metabolismo , Embarazo , Células del Estroma/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
18.
Hypertension ; 74(4): 967-974, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31378106

RESUMEN

The pregnancy-augmented uterine vasodilation is linked to increased AT2R (angiotensin type-2 receptor) that mediates the vasodilatory effects of angiotensin II. However, the mechanisms controlling AT2R expression during pregnancy remain unclear. Estrogens are known to play a role in vascular adaptations during pregnancy. We hypothesized that estrogen stimulates uterine artery AT2R expression via ER (estrogen receptor)-ß-dependent transcription in a pregnancy-specific endothelium-dependent manner. Plasma estradiol levels increased and peaked in late pregnancy and returned to prepregnant levels post-partum, correlating with uterine artery AT2R and ERß upregulation. Estradiol stimulated AT2R mRNA expression in endothelium-intact but not endothelium-denuded late pregnant and nonpregnant rat uterine artery ex vivo. Consistently, estradiol stimulated AT2R mRNA expression in late pregnant but not nonpregnant primary human uterine artery endothelial cells in vitro, which was abolished by ER antagonist ICI 182,780. Higher ERα protein bound to ER-responsive elements in AT2R promoter in the nonpregnant arteries whereas higher ERß bound in the pregnant state. ERα protein levels were similar but higher ERß protein levels were expressed in pregnant versus nonpregnant human uterine artery endothelial cells. Estradiol stimulation recruited ERα to the AT2R promoter in the nonpregnant state and ERß to the AT2R promoter in pregnancy; however, only ERß recruitment mediated transactivation of the AT2R reporter gene in pregnant human uterine artery endothelial cells. Estradiol-induced AT2R expression was abolished by the specific ERß (not ERα) antagonist 4-[2-Phenyl-5,7-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-3-yl]phenol (PHTPP) and mimicked by the specific ERß (not ERα) agonist 2,3-bis(4-Hydroxyphenyl)-propionitrile (DPN) in pregnant human uterine artery endothelial cells in vitro. This study demonstrates a novel role of pregnancy-augmented ERß in AT2R upregulation in the uterine artery and provides new insights into the mechanisms underlying uterine vascular adaptation to pregnancy.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Estradiol/farmacología , Receptor beta de Estrógeno/metabolismo , Receptor de Angiotensina Tipo 2/metabolismo , Arteria Uterina/efectos de los fármacos , Animales , Células Endoteliales/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Estradiol/sangre , Femenino , Embarazo , Ratas , Regulación hacia Arriba/efectos de los fármacos , Arteria Uterina/metabolismo
19.
Curr Med Sci ; 38(3): 513-518, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30074220

RESUMEN

This study aimed to explore the outcomes of progestin-primed ovarian stimulation protocol (PPOS) in aged infertile women who failed to get pregnant in the first IVF/ICSI-ET cycles with GnRH-a long protocol. A self-controlled study was conducted to retrospectively investigate the clinical outcomes of 104 aged infertile patients who didn't get pregnant in the first IVF/ICSI-ET treatment by stimulating with GnRH-a long protocol (non-PPOS group), and underwent PPOS protocol (PPOS group) in the second cycle between January 2016 and December 2016 in the Center for Reproductive Medicine, Renmin Hospital of Wuhan University. The primary outcomes included clinical pregnancy rate of frozen-thawed embryos transfer (FET) in PPOS group, and good-quality embryo rate in both groups. The secondary outcomes were fertilization rate, egg utilization rate and cycle cancellation rate. The results showed that there were no significant differences in basal follicle stimulating hormone (bFSH), antral follicle count (AFC), duration and total dosage of gonadotropin (Gn), number of oocytes retrieved, intracytoplasmic sperm injection (ICSI) rate, fertilization rate, and cycle cancellation rate between the two groups (P>0.05). However, the oocyte utilization rate and good-quality embryo rate in PPOS group were significantly higher than those in non-PPOS group (P<0.05). By the end of April 2017,62 FET cycles were conducted in PPOS group. The clinical pregnancy rate and embryo implantation rate were 22.58% and 12.70%, respectively. In conclusion, PPOS protocol may provide better clinical outcomes by improving the oocyte utilization rate and good-quality embryo rate for aged infertile patients who failed to get pregnant in the first IVF/ICSI-ET cycles.


Asunto(s)
Fertilización In Vitro , Infertilidad Femenina/terapia , Inducción de la Ovulación , Resultado del Embarazo , Progestinas/farmacología , Inyecciones de Esperma Intracitoplasmáticas , Adulto , Femenino , Humanos , Embarazo
20.
Theriogenology ; 106: 227-236, 2018 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-29096270

RESUMEN

Sphingolipids play multiple roles in membrane structure, signal transduction, stress responses, neural development and immune reaction. The rate of de novo synthesis pathway of sphingolipids is regulated by two key enzymes, serine palmitoyltransferase (SPT), and ketoreductase (Kds). Here, we find that the mRNA levels of three subunits of the SPT holoenzyme (Sptlc1, Sptlc2, and Ssspta) are significantly up-regulated in mouse uterine stromal cells during decidualization. The expression of Kds, which reduces 3-keto-dihydrosphingosine to dihydrosphingosine, is co-localized with Sptlc1 in mouse uteri during early pregnancy. Moreover, l-Cycloserine, a specific inhibitor of SPT, can significantly decrease the weight and number of implantation sites, and impede the decidualization process in mouse uterine stromal cells, suggesting that blockage of de novo sphingolipid synthesis may cause defective decidualization and early pregnancy loss in mice. In addition, this study also shows progesterone (P4) can stimulate the expression of both Sptlc2 and Ssspta in mouse uterus. Therefore, our study shows that de novo synthesis of sphingolipids is necessary in implantation and plays a key role in decidualization of mouse.


Asunto(s)
Decidua/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Esfingolípidos/metabolismo , Aborto Veterinario/genética , Animales , Implantación del Embrión/genética , Femenino , Ratones , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA